1
|
Zhao X, Qiao X, Yu S, Jin Y, Niu J, Li J, Xu Y, Yang Y, Wang L, Song L. KDM4 Regulates the Glycolysis of Hemocytes in the Immune Priming of Eriocheir sinensis. Int J Mol Sci 2024; 25:13174. [PMID: 39684884 DOI: 10.3390/ijms252313174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Immune priming confers a sustained, augmented response of innate immune cells to a secondary challenge, a process that is characteristically reliant on metabolic reprogramming. Recent evidence suggests that histone demethylases play essential roles in the immune priming, while its regulation role in the metabolic reprogramming remains largely unknown. In the present study, the concentration of glucose was significantly down-regulated in the hemocytes of crab Eriocheir sinensis after secondary stimulation with Aeromonas hydrophila, while the expression levels of phosphofructokinase (EsPFK) pyruvate kinase (EsPK), hexokinase-2 (EsHK-2) and Glucose-6-phosphate dehydrogenase (EsG-6-PD), along with the concentrations of lactate and the ratio of NAD+/NADH, were elevated. Additionally, the levels of H3K9me3 and its enrichment at the promoters of EsPFK and EsG-6-PD were significantly decreased at 7 days after A. hydrophila stimulation. The lysine Demethylase 4 homologue (EsKDM4) was observed to translocate into the nucleus of crab hemocytes after A. hydrophila stimulation, and its activity markedly increased after secondary stimulation with A. hydrophila. Following RNA interference of EsKDM4, there was a significant increase in H3K9me3 levels, and the enrichment of H3K9me3 at the EsPFK and EsG-6-PD promoters, as well as the concentration of glucose, in the hemocytes of crabs after secondary stimulation with A. hydrophila. Furthermore, mRNA transcripts of EsPFK and EsG-6-PD, as well as the concentration of lactate and ratio of NAD+/NADH, significantly decreased after secondary stimulation. These results suggested that EsKDM4 mediates the enrichment of H3K9me3 at the promoters of EsPFK and EsG-6-PD, thereby regulating glycolysis during the immune priming of crabs.
Collapse
Affiliation(s)
- Xinyu Zhao
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Xue Qiao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Simiao Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Yuhao Jin
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Jixiang Niu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Jie Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Yingmei Xu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Yuehong Yang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Linsheng Song
- College of Life Sciences, Liaoning Normal University, Dalian 116029, China
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
2
|
Kim J, Choi J. Histone Methylation-Mediated Reproductive Toxicity to Consumer Product Chemicals in Caenorhabditis elegans: An Epigenetic Adverse Outcome Pathway (AOP). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:19604-19616. [PMID: 39445662 PMCID: PMC11542887 DOI: 10.1021/acs.est.4c04061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
The significance of histone methylation in epigenetic inheritance underscores its relevance to disease and the chronic effects of environmental chemicals. However, limited evidence of the causal relationships between chemically induced epigenetic changes and organismal-level effects hinders the application of epigenetic markers in ecotoxicological assessments. This study explored the contribution of repressive histone marks to reproductive toxicity induced by chemicals in consumer products in Caenorhabditis elegans, applying the adverse outcome pathway (AOP) framework. Triclosan (TCS) and tetrabromobisphenol A (TBBPA) exposures caused reproductive toxicity and altered histone methyltransferase (HMT) and histone demethylase (HDM) activities, increasing the level of trimethylation of H3K9 and H3K27. Notably, treatment with an H3K27-specific HMT inhibitor alleviated reproductive defects and the transcriptional response of genes related to vitellogenin, xenobiotic metabolism, and oxidative stress. Comparison of points of departure (PODs) based on calculated benchmark concentrations (BMCs) revealed the sensitivity of histone-modifying enzyme activities to these chemicals. Our findings suggest that the 'disturbance of HMT and HDM' can serve as the molecular initiating event (MIE) leading to reproductive toxicity in the epigenetic AOP for TCS and TBBPA. The study extended the biological applicability of these enzymes by identifying model species with analogous protein sequences and functions. This combined approach enhances the essentiality, empirical support, and taxonomic domain of applicability (tDOA), which are crucial considerations for ecotoxicological AOPs. Given the widespread use and environmental distribution of chemicals in consumer products, this study proposes histone-modifying enzyme activity as an effective screening tool for reproductive toxicants and emphasizes the integration of epigenetic mechanisms into a prospective ERA.
Collapse
Affiliation(s)
- Jiwan Kim
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic
of Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic
of Korea
| |
Collapse
|
3
|
Huang X, Ye Q, Dai W, Zheng J, Li Y, Wang C, Luo Z, Yang J, Zhuo W, Wan QL. Cadmium exposure induces multigenerational inheritance of germ cell apoptosis and fertility suppression in Caenorhabditis elegans. ENVIRONMENT INTERNATIONAL 2024; 191:108952. [PMID: 39159515 DOI: 10.1016/j.envint.2024.108952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
Cadmium is a significant environmental pollutant that poses a substantial health hazard to humans due to its propensity to accumulate in the body and resist excretion. We have a comprehensive understanding of the damage caused by Cd exposure and the mechanisms of tolerance, however, the intricate mechanisms underlying multigenerational effects resulting from Cd exposure remain poorly understood. In this study, Caenorhabditis elegans were used as a model organism to investigate Cd-induced multigenerational effects and its association with epigenetic modifications. The results showed that Cd exposure leads to an increase in germ cell apoptosis and a decrease in fertility, which can be passed down to subsequent generations. Further analysis revealed that transcription factors DAF-16/FOXO and SKN-1/Nrf2 play essential roles in responding to Cd exposure and in the transgenerational induction of germ cell apoptosis. Additionally, histone H3K4 trimethylation (H3K4me3) marks stress-responsive genes and enhances their transcription, ultimately triggering multigenerational germ cell apoptosis. This study provides compelling evidence that the detrimental effects of Cd on the reproductive system can be inherited across generations. These findings enhance our understanding of the multigenerational effects of environmental pollutants and may inform strategies for the prevention and control of such pollutants.
Collapse
Affiliation(s)
- Xiaobing Huang
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China; Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai 519000, Guangdong, China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Qunshan Ye
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Wenyu Dai
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China; Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jingming Zheng
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yimin Li
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Cheng Wang
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Zhenhuan Luo
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jing Yang
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China; Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong, China
| | - Wenyan Zhuo
- Department of Neurology, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai 519000, Guangdong, China
| | - Qin-Li Wan
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
4
|
Webster AK, Phillips PC. Heritable epigenetic variation facilitates long-term maintenance of epigenetic and genetic variation. G3 (BETHESDA, MD.) 2024; 14:jkad287. [PMID: 38113034 PMCID: PMC10849368 DOI: 10.1093/g3journal/jkad287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/03/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023]
Abstract
How genetic and phenotypic variation are maintained has long been one of the fundamental questions in population and quantitative genetics. A variety of factors have been implicated to explain the maintenance of genetic variation in some contexts (e.g. balancing selection), but the potential role of epigenetic regulation to influence population dynamics has been understudied. It is well recognized that epigenetic regulation, including histone methylation, small RNA expression, and DNA methylation, helps to define differences between cell types and facilitate phenotypic plasticity. In recent years, empirical studies have shown the potential for epigenetic regulation to also be heritable for at least a few generations without selection, raising the possibility that differences in epigenetic regulation can act alongside genetic variation to shape evolutionary trajectories. Heritable differences in epigenetic regulation that arise spontaneously are termed "epimutations." Epimutations differ from genetic mutations in 2 key ways-they occur at a higher rate and the loci at which they occur often revert back to their original state within a few generations. Here, we present an extension of the standard population genetic model with selection to incorporate epigenetic variation arising via epimutation. Our model assumes a diploid, sexually reproducing population with random mating. In addition to spontaneous genetic mutation, we included parameters for spontaneous epimutation and back-epimutation, allowing for 4 potential epialleles at a single locus (2 genetic alleles, each with 2 epigenetic states), each of which affect fitness. We then analyzed the conditions under which stable epialleles were maintained. Our results show that highly reversible epialleles can be maintained in long-term equilibrium under neutral conditions in a manner that depends on the epimutation and back-epimutation rates, which we term epimutation-back-epimutation equilibrium. On the other hand, epialleles that compensate for deleterious mutations cause deviations from the expectations of mutation-selection balance by a simple factor that depends on the epimutation and back-epimutation rates. We also numerically analyze several sets of fitness parameters for which large deviations from mutation-selection balance occur. Together, these results demonstrate that transient epigenetic regulation may be an important factor in the maintenance of both epigenetic and genetic variation in populations.
Collapse
Affiliation(s)
- Amy K Webster
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Patrick C Phillips
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
5
|
Dias BG. Legacies of salient environmental experiences-insights from chemosensation. Chem Senses 2024; 49:bjae002. [PMID: 38219073 PMCID: PMC10825851 DOI: 10.1093/chemse/bjae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Indexed: 01/15/2024] Open
Abstract
Evidence for parental environments profoundly influencing the physiology, biology, and neurobiology of future generations has been accumulating in the literature. Recent efforts to understand this phenomenon and its underlying mechanisms have sought to use species like rodents and insects to model multi-generational legacies of parental experiences like stress and nutritional exposures. From these studies, we have come to appreciate that parental exposure to salient environmental experiences impacts the cadence of brain development, hormonal responses to stress, and the expression of genes that govern cellular responses to stress in offspring. Recent studies using chemosensory exposure have emerged as a powerful tool to shed new light on how future generations come to be influenced by environments to which parents are exposed. With a specific focus on studies that have leveraged such use of salient chemosensory experiences, this review synthesizes our current understanding of the concept, causes, and consequences of the inheritance of chemosensory legacies by future generations and how this field of inquiry informs the larger picture of how parental experiences can influence offspring biology.
Collapse
Affiliation(s)
- Brian G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, United States
- Division of Endocrinology, Diabetes and Metabolism, Children’s Hospital Los Angeles, Los Angeles, CA, United States
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, United States
| |
Collapse
|
6
|
Chen H, Gu Y, Jiang Y, Yu J, Chen C, Shi C, Li H. Photoaged Polystyrene Nanoplastics Result in Transgenerational Reproductive Toxicity Associated with the Methylation of Histone H3K4 and H3K9 in Caenorhabditis elegans. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:19341-19351. [PMID: 37934861 DOI: 10.1021/acs.est.3c05861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Polystyrene nanoplastics (PS-NPs) are emerging environmental contaminants that are ubiquitously detected in various environments and have toxic effects on various organisms. Nevertheless, the transgenerational reproductive toxicity and underlying mechanisms of PS-NPs remain largely unknown, especially for photoaged PS-NPs under ultraviolet irradiation. In this study, only the parental generation (P0) was exposed to virgin and aged PS-NPs at environmentally relevant concentrations (0.1-100 μg/L), and subsequent generations (F1-F4) were cultured under normal conditions. Ultraviolet irradiation induced the generation of environmentally persistent free radicals and reactive oxygen species, which altered the physical and chemical characteristics of PS-NPs. The results of toxicity testing suggested that exposure to aged PS-NPs caused a more severe decrease in brood size, egg ejection rate, number of fertilized eggs, and hatchability than did the virgin PS-NPs in the P0, F1, and F2 generations. Additionally, a single maternal exposure to aged PS-NPs resulted in transgenerational effects on fertility in the F1 and F2 generations. Increased levels of H3K4 and H3K9 methylation were observed in the F1 and F2 generations, which were concomitant with the transgenerational downregulation of the expression of associated genes, such as spr-5, set-17, and met-2. On the basis of correlation analyses, the levels of histone methylation and the expression of these genes were significantly correlated to transgenerational reproductive effects. Further research showed that transgenerational effects on fertility were not observed in spr-5(by134), met-2(n4256), and set-17(n5017) mutants. Overall, maternal exposure to aged PS-NPs induced transgenerational reproductive effects via H3K4 and H3K9 methylation, and the spr-5, met-2, and set-17 genes were involved in the regulation of transgenerational toxicity. This study provides new insights into the potential risks of photoaging PS-NPs in the environment.
Collapse
Affiliation(s)
- Haibo Chen
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yulun Gu
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yongqi Jiang
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Jun Yu
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Chao Chen
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Chongli Shi
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Hui Li
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
7
|
Blumberg B, Cheng-An Chang R, Egusquiza R, Amato A, Li Z, Joloya E, Wheeler H, Nguyen A, Shioda K, Odajima J, Lawrence M, Shioda T. Heritable changes in chromatin contacts linked to transgenerational obesity. RESEARCH SQUARE 2023:rs.3.rs-3570919. [PMID: 38077066 PMCID: PMC10705594 DOI: 10.21203/rs.3.rs-3570919/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Burgeoning evidence demonstrates that effects of environmental exposures can be transmitted to subsequent generations through the germline without DNA mutations1,2. This phenomenon remains controversial because underlying mechanisms have not been identified. Therefore, understanding how effects of environmental exposures are transmitted to unexposed generations without DNA mutations is a fundamental unanswered question in biology. Here, we used an established murine model of male-specific transgenerational obesity to show that exposure to the obesogen tributyltin (TBT) elicited heritable changes in chromatin interactions (CIs) in primordial germ cells (PGCs). New CIs were formed within the Ide gene encoding Insulin Degrading Enzyme in the directly exposed PGCs, then stably maintained in PGCs of the subsequent (unexposed) two generations. Concomitantly, Ide mRNA expression was decreased in livers of male descendants from the exposed dams. These males were hyperinsulinemic and hyperglycemic, phenocopying Ide-deficient mice that are predisposed to adult-onset, diet-induced obesity. Creation of new CIs in PGCs, suppression of hepatic Ide mRNA, increased fat mass, hyperinsulinemia and hyperglycemia were male-specific. Our results provide a plausible molecular mechanism underlying transmission of the transgenerational predisposition to obesity caused by gestational exposure to an environmental obesogen. They also provide an entry point for future studies aimed at understanding how environmental exposures alter chromatin structure to influence physiology across multiple generations in mammals.
Collapse
|
8
|
Yuan M, Zeng C, Lu H, Yue Y, Sun T, Zhou X, Li G, Ai N, Ge W. Genetic and Epigenetic Evidence for Nonestrogenic Disruption of Otolith Development by Bisphenol A in Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:16190-16205. [PMID: 37752410 DOI: 10.1021/acs.est.3c04336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Bisphenol A (BPA) is a well-known endocrine-disrupting chemical (EDC) that has estrogenic activities. In addition to disrupting reproductive development and function via estrogenic signaling pathways, BPA can also interfere with nonreproductive functions through nonestrogenic pathways; however, the mechanisms underlying such nonestrogenic activities are not well understood. In this study, we demonstrated that BPA could disrupt otolith formation during the early development of zebrafish with long-lasting ethological effects. Using multiple mutants of estrogen receptors, we provided strong genetic evidence that the BPA-induced otolith malformation was independent of estrogen signaling. Transcriptome analysis revealed that two genes related to otolith development, otopetrin 1 (otop1) and starmaker (stm), decreased their expression significantly after BPA exposure. Knockout of both otop1 and stm genes could phenocopy the BPA-induced otolith malformation, while microinjection of their mRNAs could rescue the BPA-induced abnormalities of otolith formation. Further experiments showed that BPA inhibited the expression of otop1 and stm by activating the MEK/ERK-EZH2-H3K27me3 signaling pathway. Taken together, our study provided comprehensive genetic and molecular evidence that BPA induced the otolith malformation through nonestrogenic pathway during zebrafish early development and its activities involved epigenetic control of key genes (e.g., otop1 and stm) participating in otolith formation.
Collapse
Affiliation(s)
- Mingzhe Yuan
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Chu Zeng
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Huijie Lu
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Yiming Yue
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Ting Sun
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Gang Li
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Nana Ai
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| | - Wei Ge
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa 999078, Macau, China
| |
Collapse
|
9
|
Peng JY, Liu X, Zeng XT, Hao Y, Zhang JH, Li Q, Tong XJ. Early pheromone perception remodels neurodevelopment and accelerates neurodegeneration in adult C. elegans. Cell Rep 2023; 42:112598. [PMID: 37289584 DOI: 10.1016/j.celrep.2023.112598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
Age-associated neurodegenerative disorders such as Parkinson's and Alzheimer's diseases are mainly caused by protein aggregation. The etiologies of these neurodegenerative diseases share a chemical environment. However, how chemical cues modulate neurodegeneration remains unclear. Here, we found that in Caenorhabditis elegans, exposure to pheromones in the L1 stage accelerates neurodegeneration in adults. Perception of pheromones ascr#3 and ascr#10 is mediated by chemosensory neurons ASK and ASI. ascr#3 perceived by G protein-coupled receptor (GPCR) DAF-38 in ASK activates glutamatergic transmission into AIA interneurons. ascr#10 perceived by GPCR STR-2 in ASI activates the secretion of neuropeptide NLP-1, which binds to the NPR-11 receptor in AIA. Activation of both ASI and ASK is required and sufficient to remodel neurodevelopment via AIA, which triggers insulin-like signaling and inhibits autophagy in adult neurons non-cell-autonomously. Our work reveals how pheromone perception at the early developmental stage modulates neurodegeneration in adults and provides insights into how the external environment impacts neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing-Yi Peng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuqing Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xian-Ting Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yue Hao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia-Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Lingang Laboratory, Shanghai 200031, China
| | - Qian Li
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Center for Brain Science, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Department of Anatomy and Physiology, Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health in Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia-Jing Tong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
10
|
Truong L, Chen YW, Barrere-Cain R, Levenson MT, Shuck K, Xiao W, da Veiga Beltrame E, Panter B, Reich E, Sternberg PW, Yang X, Allard P. Single-nucleus resolution mapping of the adult C. elegans and its application to elucidate inter- and trans-generational response to alcohol. Cell Rep 2023; 42:112535. [PMID: 37227821 PMCID: PMC10592506 DOI: 10.1016/j.celrep.2023.112535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 03/16/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Single-cell transcriptomic platforms provide an opportunity to map an organism's response to environmental cues with high resolution. Here, we applied single-nucleus RNA sequencing (snRNA-seq) to establish the tissue and cell type-resolved transcriptome of the adult C. elegans and characterize the inter- and trans-generational transcriptional impact of ethanol. We profiled the transcriptome of 41,749 nuclei resolving into 31 clusters, representing a diverse array of adult cell types including syncytial tissues. Following exposure to human-relevant doses of alcohol, several germline, striated muscle, and neuronal clusters were identified as being the most transcriptionally impacted at the F1 and F3 generations. The effect on germline clusters was confirmed by phenotypic enrichment analysis as well as by functional validation, which revealed a remarkable inter- and trans-generational increase in germline apoptosis, aneuploidy, and embryonic lethality. Together, snRNA-seq represents a valuable approach for the detailed examination of an adult organism's response to environmental exposures.
Collapse
Affiliation(s)
- Lisa Truong
- Human Genetics Graduate Program, UCLA, Los Angeles, CA 90095, USA
| | - Yen-Wei Chen
- Molecular Toxicology Inter-Departmental Program, UCLA, Los Angeles, CA 90095, USA
| | - Rio Barrere-Cain
- Institute for Society & Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Max T Levenson
- Molecular Toxicology Inter-Departmental Program, UCLA, Los Angeles, CA 90095, USA
| | - Karissa Shuck
- Institute for Society & Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Wen Xiao
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | | | - Blake Panter
- Institute for Society & Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Ella Reich
- Institute for Society & Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Paul W Sternberg
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Xia Yang
- Integrative Biology and Physiology Department, UCLA, Los Angeles, CA 90095, USA
| | - Patrick Allard
- Molecular Toxicology Inter-Departmental Program, UCLA, Los Angeles, CA 90095, USA; Institute for Society & Genetics, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Abstract
After decades of research, our knowledge of the complexity of cancer mechanisms, elegantly summarized as 'hallmarks of cancer', is expanding, as are the therapeutic opportunities that this knowledge brings. However, cancer still needs intense research to diminish its tremendous impact. In this context, the use of simple model organisms such as Caenorhabditis elegans, in which the genetics of the apoptotic pathway was discovered, can facilitate the investigation of several cancer hallmarks. Amenable for genetic and drug screens, convenient for fast and efficient genome editing, and aligned with the 3Rs ('Replacement, Reduction and Refinement') principles for ethical animal research, C. elegans plays a significant role in unravelling the intricate network of cancer mechanisms and presents a promising option in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Julián Cerón
- Modeling Human Diseases in C. elegans Group – Genes, Disease and Therapy Program, Bellvitge Biomedical Research Institute – IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
12
|
Camacho JA, Welch B, Sprando RL, Hunt PR. Reproductive-Toxicity-Related Endpoints in C. elegans Are Consistent with Reduced Concern for Dimethylarsinic Acid Exposure Relative to Inorganic Arsenic. J Dev Biol 2023; 11:18. [PMID: 37218812 PMCID: PMC10204422 DOI: 10.3390/jdb11020018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Exposures to arsenic and mercury are known to pose significant threats to human health; however, the effects specific to organic vs. inorganic forms are not fully understood. Caenorhabditis elegans' (C. elegans) transparent cuticle, along with the conservation of key genetic pathways regulating developmental and reproductive toxicology (DART)-related processes such as germ stem cell renewal and differentiation, meiosis, and embryonic tissue differentiation and growth, support this model's potential to address the need for quicker and more dependable testing methods for DART hazard identification. Organic and inorganic forms of mercury and arsenic had different effects on reproductive-related endpoints in C. elegans, with methylmercury (meHgCl) having effects at lower concentrations than mercury chloride (HgCl2), and sodium arsenite (NaAsO2) having effects at lower concentrations than dimethylarsinic acid (DMA). Progeny to adult ratio changes and germline apoptosis were seen at concentrations that also affected gravid adult gross morphology. For both forms of arsenic tested, germline histone regulation was altered at concentrations below those that affected progeny/adult ratios, while concentrations for these two endpoints were similar for the mercury compounds. These C. elegans findings are consistent with corresponding mammalian data, where available, suggesting that small animal model test systems may help to fill critical data gaps by contributing to weight of evidence assessments.
Collapse
Affiliation(s)
- Jessica A. Camacho
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA
| | | | | | | |
Collapse
|
13
|
D’Cruz SC, Hao C, Labussiere M, Mustieles V, Freire C, Legoff L, Magnaghi-Jaulin L, Olivas-Martinez A, Rodriguez-Carrillo A, Jaulin C, David A, Fernandez MF, Smagulova F. Genome-wide distribution of histone trimethylation reveals a global impact of bisphenol A on telomeric binding proteins and histone acetyltransferase factors: a pilot study with human and in vitro data. Clin Epigenetics 2022; 14:186. [PMID: 36572933 PMCID: PMC9793539 DOI: 10.1186/s13148-022-01408-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/14/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To assess the genetic and epigenetic effects promoted by Bisphenol A (BPA) exposure in adolescent males from the Spanish INMA-Granada birth cohort, and in human cells. METHODS DNA methylation was analysed using MEDIP. Repeat number variation in genomic DNA was evaluated, along with the analysis of H3K4me3 by using chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq). Analyses were performed with material extracted from whole blood of the adolescents, complemented by in vitro assessments of human (HeLa) cells exposed to 10 nM BPA, specifically, immunofluorescence evaluation of protein levels, gene expression analysis and ChIP‒qPCR analysis. RESULTS Adolescents in the high urinary BPA levels group presented a higher level of Satellite A (SATA) repetitive region copy numbers compared to those in the low BPA group and a tendency towards increase in telomere length. We also observed decreased DNA methylation at the promoters of the imprinted genes H19, KCNQ1, and IGF2; at LINE1 retroelements; and at the ARID2, EGFR and ESRRA and TERT genes. Genome-wide sequencing revealed increased H3K4me3 occupancy at the promoters of genes encoding histone acetyltransferases, telomeric DNA binding factors and DNA repair genes. Results were supported in HeLa cells exposed to 10 nM BPA in vitro. In accordance with the data obtained in blood samples, we observed higher H3K4me3 occupancy and lower DNA methylation at some specific targets in HeLa cells. In exposed cells, changes in the expression of genes encoding DNA repair factors (ATM, ARID2, TRP53) were observed, and increased expression of several genes encoding telomeric DNA binding factors (SMG7, TERT, TEN1, UPF1, ZBTB48) were also found. Furthermore, an increase in ESR1/ERa was observed in the nuclei of HeLa cells along with increased binding of ESR1 to KAT5, KMT2E and TERF2IP promoters and decreased ESR1 binding at the RARA promoter. The DNA damage marker p53/TP53 was also increased. CONCLUSION In this pilot study, genome-wide analysis of histone trimethylation in adolescent males exposed to BPA revealed a global impact on the expression of genes encoding telomeric binding proteins and histone acetyltransferase factors with similar results in HeLa cells. Nevertheless, larger studies should confirm our findings.
Collapse
Affiliation(s)
- Shereen Cynthia D’Cruz
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| | - Chunxiang Hao
- grid.410747.10000 0004 1763 3680School of Medicine, Linyi University, Linyi, 276000 China
| | - Martin Labussiere
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| | - Vicente Mustieles
- grid.4489.10000000121678994Center for Biomedical Research (CIBM), Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016 Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria (Ibs.GRANADA), 18012 Granada, Spain ,grid.466571.70000 0004 1756 6246Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Carmen Freire
- grid.4489.10000000121678994Center for Biomedical Research (CIBM), Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016 Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria (Ibs.GRANADA), 18012 Granada, Spain ,grid.466571.70000 0004 1756 6246Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Louis Legoff
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| | - Laura Magnaghi-Jaulin
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| | - Alicia Olivas-Martinez
- grid.4489.10000000121678994Center for Biomedical Research (CIBM), Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016 Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria (Ibs.GRANADA), 18012 Granada, Spain
| | - Andrea Rodriguez-Carrillo
- grid.4489.10000000121678994Center for Biomedical Research (CIBM), Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016 Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria (Ibs.GRANADA), 18012 Granada, Spain
| | - Christian Jaulin
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| | - Arthur David
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| | - Mariana F. Fernandez
- grid.4489.10000000121678994Center for Biomedical Research (CIBM), Department of Radiology and Physical Medicine, School of Medicine, University of Granada, 18016 Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria (Ibs.GRANADA), 18012 Granada, Spain ,grid.466571.70000 0004 1756 6246Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Fatima Smagulova
- grid.410368.80000 0001 2191 9284EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, University Rennes, 35000 Rennes, France
| |
Collapse
|
14
|
Sun B, Sherrin M, Roy R. Unscheduled epigenetic modifications cause genome instability and sterility through aberrant R-loops following starvation. Nucleic Acids Res 2022; 51:84-98. [PMID: 36504323 PMCID: PMC9841415 DOI: 10.1093/nar/gkac1155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
During starvation, organisms modify both gene expression and metabolism to adjust to the energy stress. We previously reported that Caenorhabditis elegans lacing AMP-activated protein kinase (AMPK) exhibit transgenerational reproductive defects associated with abnormally elevated trimethylated histone H3 at lysine 4 (H3K4me3) levels in the germ line following recovery from acute starvation. Here, we show that these H3K4me3 marks are significantly increased at promoters, driving aberrant transcription elongation resulting in the accumulation of R-loops in starved AMPK mutants. DNA-RNA immunoprecipitation followed by high-throughput sequencing (DRIP-seq) analysis demonstrated that a significant proportion of the genome was affected by R-loop formation. This was most pronounced in the promoter-transcription start site regions of genes, in which the chromatin was modified by H3K4me3. Like H3K4me3, the R-loops were also found to be heritable, likely contributing to the transgenerational reproductive defects typical of these mutants following starvation. Strikingly, AMPK mutant germ lines show considerably more RAD-51 (the RecA recombinase) foci at sites of R-loop formation, potentially sequestering them from their roles at meiotic breaks or at sites of induced DNA damage. Our study reveals a previously unforeseen role of AMPK in maintaining genome stability following starvation. The downstream effects of R-loops on DNA damage sensitivity and germline stem cell integrity may account for inappropriate epigenetic modification that occurs in numerous human disorders, including various cancers.
Collapse
Affiliation(s)
- Bing Sun
- To whom correspondence should be addressed.
| | - McLean Sherrin
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Richard Roy
- Correspondence may also be addressed to Richard Roy. Tel: +1 514 398 6437;
| |
Collapse
|
15
|
Li H, Zeng L, Wang C, Shi C, Li Y, Peng Y, Chen H, Zhang J, Cheng B, Chen C, Xiang M, Huang Y. Review of the toxicity and potential molecular mechanisms of parental or successive exposure to environmental pollutants in the model organism Caenorhabditis elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 311:119927. [PMID: 35970344 DOI: 10.1016/j.envpol.2022.119927] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Environmental pollutants such as heavy metals, nano/microparticles, and organic compounds have been detected in a wide range of environmental media, causing long-term exposure in various organisms and even humans through breathing, contacting, ingestion, and other routes. Long-term exposure to environmental pollutants in organisms or humans promotes exposure of offspring to parental and environmental pollutants, and subsequently results in multiple biological defects in the offspring. This review dialectically summarizes and discusses the existing studies using Caenorhabditis elegans (C. elegans) as a model organism to explore the multi/transgenerational toxicity and potential underlying molecular mechanisms induced by environmental pollutants following parental or successive exposure patterns. Parental and successive exposure to environmental pollutants induces various biological defects in C. elegans across multiple generations, including multi/transgenerational developmental toxicity, neurotoxicity, reproductive toxicity, and metabolic disturbances, which may be transmitted to progeny through reactive oxygen species-induced damage, epigenetic mechanisms, insulin/insulin-like growth factor-1 signaling pathway. This review aims to arouse researchers' interest in the multi/transgenerational toxicity of pollutants and hopes to explore the possible long-term effects of environmental pollutants on organisms and even humans, as well as to provide constructive suggestions for the safety and management of emerging alternatives.
Collapse
Affiliation(s)
- Hui Li
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Lingjun Zeng
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Chen Wang
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| | - Chongli Shi
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Yeyong Li
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Yi Peng
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Haibo Chen
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou, 510655, China
| | - Jin Zhang
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Biao Cheng
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Chao Chen
- State Key Laboratory of Bioreactor Engineering, Biomedical Nanotechnology Center, Shanghai Collaborative Innovation Center for Biomanufacturing, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Minghui Xiang
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Yuan Huang
- Institute for Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| |
Collapse
|
16
|
Heindel JJ, Howard S, Agay-Shay K, Arrebola JP, Audouze K, Babin PJ, Barouki R, Bansal A, Blanc E, Cave MC, Chatterjee S, Chevalier N, Choudhury M, Collier D, Connolly L, Coumoul X, Garruti G, Gilbertson M, Hoepner LA, Holloway AC, Howell G, Kassotis CD, Kay MK, Kim MJ, Lagadic-Gossmann D, Langouet S, Legrand A, Li Z, Le Mentec H, Lind L, Monica Lind P, Lustig RH, Martin-Chouly C, Munic Kos V, Podechard N, Roepke TA, Sargis RM, Starling A, Tomlinson CR, Touma C, Vondracek J, Vom Saal F, Blumberg B. Obesity II: Establishing causal links between chemical exposures and obesity. Biochem Pharmacol 2022; 199:115015. [PMID: 35395240 PMCID: PMC9124454 DOI: 10.1016/j.bcp.2022.115015] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a multifactorial disease with both genetic and environmental components. The prevailing view is that obesity results from an imbalance between energy intake and expenditure caused by overeating and insufficient exercise. We describe another environmental element that can alter the balance between energy intake and energy expenditure: obesogens. Obesogens are a subset of environmental chemicals that act as endocrine disruptors affecting metabolic endpoints. The obesogen hypothesis posits that exposure to endocrine disruptors and other chemicals can alter the development and function of the adipose tissue, liver, pancreas, gastrointestinal tract, and brain, thus changing the set point for control of metabolism. Obesogens can determine how much food is needed to maintain homeostasis and thereby increase the susceptibility to obesity. The most sensitive time for obesogen action is in utero and early childhood, in part via epigenetic programming that can be transmitted to future generations. This review explores the evidence supporting the obesogen hypothesis and highlights knowledge gaps that have prevented widespread acceptance as a contributor to the obesity pandemic. Critically, the obesogen hypothesis changes the narrative from curing obesity to preventing obesity.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA.
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, USA
| | - Keren Agay-Shay
- Health and Environment Research (HER) Lab, The Azrieli Faculty of Medicine, Bar Ilan University, Israel
| | - Juan P Arrebola
- Department of Preventive Medicine and Public Health University of Granada, Granada, Spain
| | - Karine Audouze
- Department of Systems Biology and Bioinformatics, University of Paris, INSERM, T3S, Paris France
| | - Patrick J Babin
- Department of Life and Health Sciences, University of Bordeaux, INSERM, Pessac France
| | - Robert Barouki
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Etienne Blanc
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, USA
| | - Nicolas Chevalier
- Obstetrics and Gynecology, University of Cote d'Azur, Cote d'Azur, France
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisa Connolly
- The Institute for Global Food Security, School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Xavier Coumoul
- Department of Biochemistry, University of Paris, INSERM, T3S, 75006 Paris, France
| | - Gabriella Garruti
- Department of Endocrinology, University of Bari "Aldo Moro," Bari, Italy
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland
| | - Lori A Hoepner
- Department of Environmental and Occupational Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Alison C Holloway
- McMaster University, Department of Obstetrics and Gynecology, Hamilton, Ontario, CA, USA
| | - George Howell
- Center for Environmental Health Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Christopher D Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, USA
| | - Mathew K Kay
- College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Min Ji Kim
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | | | - Sophie Langouet
- Univ Rennes, INSERM EHESP, IRSET UMR_5S 1085, 35000 Rennes, France
| | - Antoine Legrand
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Zhuorui Li
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Helene Le Mentec
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - P Monica Lind
- Occupational and Environmental Medicine, Department of Medical Sciences, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California San Francisco, CA 94143, USA
| | | | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Normand Podechard
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Troy A Roepke
- Department of Animal Science, School of Environmental and Biological Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Robert M Sargis
- Division of Endocrinology, Diabetes and Metabolism, The University of Illinois at Chicago, Chicago, Il 60612, USA
| | - Anne Starling
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Charbel Touma
- Sorbonne Paris Nord University, Bobigny, INSERM U1124 (T3S), Paris, France
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Frederick Vom Saal
- Division of Biological Sciences, The University of Missouri, Columbia, MO 65211, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Wan QL, Meng X, Wang C, Dai W, Luo Z, Yin Z, Ju Z, Fu X, Yang J, Ye Q, Zhang ZH, Zhou Q. Histone H3K4me3 modification is a transgenerational epigenetic signal for lipid metabolism in Caenorhabditis elegans. Nat Commun 2022; 13:768. [PMID: 35140229 PMCID: PMC8828817 DOI: 10.1038/s41467-022-28469-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/20/2022] [Indexed: 11/09/2022] Open
Abstract
As a major risk factor to human health, obesity presents a massive burden to people and society. Interestingly, the obese status of parents can cause progeny's lipid accumulation through epigenetic inheritance in multiple species. To date, many questions remain as to how lipid accumulation leads to signals that are transmitted across generations. In this study, we establish a nematode model of C. elegans raised on a high-fat diet (HFD) that leads to measurable lipid accumulation, which can transmit the lipid accumulation signal to their multigenerational progeny. Using this model, we find that transcription factors DAF-16/FOXO and SBP-1/SREBP, nuclear receptors NHR-49 and NHR-80, and delta-9 desaturases (fat-5, fat-6, and fat-7) are required for transgenerational lipid accumulation. Additionally, histone H3K4 trimethylation (H3K4me3) marks lipid metabolism genes and increases their transcription response to multigenerational obesogenic effects. In summary, this study establishes an interaction between a network of lipid metabolic genes and chromatin modifications, which work together to achieve transgenerational epigenetic inheritance of obesogenic effects.
Collapse
Affiliation(s)
- Qin-Li Wan
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China.,Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiao Meng
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Chongyang Wang
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Wenyu Dai
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Zhenhuan Luo
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Zhinan Yin
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regeneration Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaodie Fu
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jing Yang
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Qunshan Ye
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China.,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Zhan-Hui Zhang
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qinghua Zhou
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan, 523560, Guangdong, China. .,The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
18
|
Ma X, Wang B, Li Z, Ding X, Wen Y, Shan W, Hu W, Wang X, Xia Y. Effects of glufosinate-ammonium on male reproductive health: Focus on epigenome and transcriptome in mouse sperm. CHEMOSPHERE 2022; 287:132395. [PMID: 34597628 DOI: 10.1016/j.chemosphere.2021.132395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/18/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
Glufosinate-ammonium (GLA) is a widely used herbicide with emerging concern over its neural and reproductive toxicity. To uncover potential effects of GLA on male reproductive health in mammals, adult male C57BL/6J mice were administered 0.2 mg/kg·d GLA for 5 weeks. After examination on fertility, testis histology and semen quality in the GLA group, we performed deep sequencing to identify repressive epigenetic marks including DNA methylation and histone modifications (H3K27me3 and H3K9me3), together with mRNA transcript levels in sperm. Then, we integrated multi-omics sequencing data to comprehensively explore GLA-induced epigenetic and transcriptomic alterations. We found no significant difference either on fertility, testis histology or semen quality-related indicators. As for epigenome, the protein level of H3K27me3 was significantly increased in GLA sperm. Next generation sequencing showed alterations of these epigenetic marks and extensive transcription inhibition in sperm. These differential repressive marks were mainly distributed at intergenic regions and introns. According to results by Gene Ontology enrichment analysis, both differentially methylated and expressed genes were mainly enriched in pathways related to synapse organization. Subtle differences in genomic imprinting were also observed between the two groups. These results suggested that GLA predominantly impaired sperm epigenome and transcriptome in mice, with little effect on fertility, testis histology or semen quality. Further studies on human sperm using similar strategies need to be conducted for a better understanding of the male reproductive toxicity of GLA.
Collapse
Affiliation(s)
- Xuan Ma
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Bingqian Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhe Li
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xingwang Ding
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ya Wen
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wenqi Shan
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Weiyue Hu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
19
|
Wang HD, Allard P. Challenging dogmas: How transgenerational epigenetics reshapes our views on life. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2022; 337:70-74. [PMID: 33900057 PMCID: PMC8546026 DOI: 10.1002/jez.2465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 01/03/2023]
Abstract
The emergence of the field of transgenerational epigenetics inheritance (TEI) has profoundly reshaped our understanding of the relationships between environment, soma, and germ cells as well as of heredity. TEI refers to the changes in chromatin state, gene expression, and/or phenotypes that are transmitted across several generations without involving changes to the DNA sequences. TEI has direct connections with, and feeds from, the fields of molecular biology, genetics, developmental biology, and reproductive biology, among others. However, the expansion of TEI-related research, has profoundly reshaped boundaries within each field and often led to the erosion of theories and concepts considered as tenets of biology. We first explore how the molecularization of biology has shifted the definition of epigenetics to include the notion of heredity and how epigenetics has refined our understanding of the central dogma of biology. The demonstrated transfer of environmental information from soma to germ cell through extracellular vesicles and subsequent alteration of health outcomes in offspring has put a definite end to the long-held principle of the Weismann barrier. TEI has also simultaneously led to the revival of the inheritance of acquired characteristics while further eroding the concept of an epigenetic "blank slate" in mammals. Using an historical framework, and via the exploration of central studies in the field, in this perspective article, we will draw a compelling argument for the revolutionary aspect of TEI in biology.
Collapse
Affiliation(s)
- Harrison D. Wang
- The Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, California. USA
| | - Patrick Allard
- The Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, California. USA
- Molecular Biology Institute, University of Los Angeles, California. Los Angeles, California. USA
| |
Collapse
|
20
|
Mohajer N, Joloya EM, Seo J, Shioda T, Blumberg B. Epigenetic Transgenerational Inheritance of the Effects of Obesogen Exposure. Front Endocrinol (Lausanne) 2021; 12:787580. [PMID: 34975759 PMCID: PMC8716683 DOI: 10.3389/fendo.2021.787580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and metabolic disorders have become a worldwide pandemic affecting millions of people. Although obesity is a multifaceted disease, there is growing evidence supporting the obesogen hypothesis, which proposes that exposure to a subset of endocrine disrupting chemicals (EDCs), known as obesogens, promotes obesity. While these effects can be observed in vitro using cell models, in vivo and human epidemiological studies have strengthened this hypothesis. Evidence from animal models showed that the effects of obesogen exposure can be inherited transgenerationally through at least the F4 generation. Transgenerational effects of EDC exposure predispose future generations to undesirable phenotypic traits and diseases, including obesity and related metabolic disorders. The exact mechanisms through which phenotypic traits are passed from an exposed organism to their offspring, without altering the primary DNA sequence, remain largely unknown. Recent research has provided strong evidence suggesting that a variety of epigenetic mechanisms may underlie transgenerational inheritance. These include differential DNA methylation, histone methylation, histone retention, the expression and/or deposition of non-coding RNAs and large-scale alterations in chromatin structure and organization. This review highlights the most recent advances in the field of epigenetics with respect to the transgenerational effects of environmental obesogens. We highlight throughout the paper the strengths and weaknesses of the evidence for proposed mechanisms underlying transgenerational inheritance and why none of these is sufficient to fully explain the phenomenon. We propose that changes in higher order chromatin organization and structure may be a plausible explanation for how some disease predispositions are heritable through multiple generations, including those that were not exposed. A solid understanding of these possible mechanisms is essential to fully understanding how environmental exposures can lead to inherited susceptibility to diseases such as obesity.
Collapse
Affiliation(s)
- Nicole Mohajer
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, United States
| | - Erika M. Joloya
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Jeongbin Seo
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Toshi Shioda
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, United States
| | - Bruce Blumberg
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, United States
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, CA, United States
| |
Collapse
|
21
|
Das S, Min S, Prahlad V. Gene bookmarking by the heat shock transcription factor programs the insulin-like signaling pathway. Mol Cell 2021; 81:4843-4860.e8. [PMID: 34648748 PMCID: PMC8642288 DOI: 10.1016/j.molcel.2021.09.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/09/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
Maternal stress can have long-lasting epigenetic effects on offspring. To examine how epigenetic changes are triggered by stress, we examined the effects of activating the universal stress-responsive heat shock transcription factor HSF-1 in the germline of Caenorhabditis elegans. We show that, when activated in germ cells, HSF-1 recruits MET-2, the putative histone 3 lysine 9 (H3K9) methyltransferase responsible for repressive H3K9me2 (H3K9 dimethyl) marks in chromatin, and negatively bookmarks the insulin receptor daf-2 and other HSF-1 target genes. Increased H3K9me2 at these genes persists in adult progeny and shifts their stress response strategy away from inducible chaperone expression as a mechanism to survive stress and instead rely on decreased insulin/insulin growth factor (IGF-1)-like signaling (IIS). Depending on the duration of maternal heat stress exposure, this epigenetic memory is inherited by the next generation. Thus, paradoxically, HSF-1 recruits the germline machinery normally responsible for erasing transcriptional memory but, instead, establishes a heritable epigenetic memory of prior stress exposure.
Collapse
Affiliation(s)
- Srijit Das
- Department of Biology, Aging Mind and Brain Initiative, 143 Biology Building, Iowa City, IA 52242-1324, USA
| | - Sehee Min
- Department of Biology, Aging Mind and Brain Initiative, 143 Biology Building, Iowa City, IA 52242-1324, USA
| | - Veena Prahlad
- Department of Biology, Aging Mind and Brain Initiative, 143 Biology Building, Iowa City, IA 52242-1324, USA; Department of Biology, 143 Biology Building, Iowa City, IA 52242-1324, USA; Iowa Neuroscience Institute, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA 52242, USA.
| |
Collapse
|
22
|
Šrut M. Ecotoxicological epigenetics in invertebrates: Emerging tool for the evaluation of present and past pollution burden. CHEMOSPHERE 2021; 282:131026. [PMID: 34111635 DOI: 10.1016/j.chemosphere.2021.131026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 06/12/2023]
Abstract
The effect of environmental pollution on epigenetic changes and their heredity in affected organisms is of major concern as such changes can play a significant role in adaptation to changing environmental conditions. Changes of epigenetic marks including DNA methylation, histone modifications, and non-coding RNA's can induce changes in gene transcription leading to physiological long-term changes or even transgenerational inheritance. Such mechanisms have until recently been scarcely studied in invertebrate organisms, mainly focusing on model species including Caenorhabditis elegans and Daphnia magna. However, more data are becoming available, particularly focused on DNA methylation changes caused by anthropogenic pollutants in a wide range of invertebrates. This review examines the literature from field and laboratory studies utilising invertebrate species exposed to environmental pollutants and their effect on DNA methylation. Possible mechanisms of epigenetic modifications and their role on physiology and adaptation as well as the incidence of intergenerational and transgenerational inheritance are discussed. Furthermore, critical research challenges are defined and the way forward is proposed. Future studies should focus on the use of next generation sequencing tools to define invertebrate methylomes under environmental stress in higher resolution, those data should further be linked to gene expression patterns and phenotypes and detailed studies focusing on transgenerational effects are encouraged. Moreover, studies of other epigenetic mechanisms in various invertebrate species, apart from DNA methylation would provide better understanding of interconnected cross-talk between epigenetic marks. Taken together incorporating epigenetic studies in ecotoxicology context presents a promising tool for development of sensitive biomarkers for environmental stress assessment.
Collapse
Affiliation(s)
- Maja Šrut
- University of Innsbruck, Institute of Zoology, Technikerstraße 25, 6020, Innsbruck, Austria.
| |
Collapse
|
23
|
Jiang X, Zhu X, Cheng Y, Azhar M, Xing X, Li W, Cao Y, Shi Q, Bao J. The Spin1 interactor, Spindoc, is dispensable for meiotic division, but essential for haploid spermatid development in mice. Reprod Biol Endocrinol 2021; 19:144. [PMID: 34526015 PMCID: PMC8442334 DOI: 10.1186/s12958-021-00828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/03/2021] [Indexed: 11/21/2022] Open
Abstract
In mammals, germline development undergoes dramatic morphological and molecular changes and is epigenetically subject to intricate yet exquisite regulation. Which epigenetic players and how they participate in the germline developmental process are not fully characterized. Spin1 is a multifunctional epigenetic protein reader that has been shown to recognize H3 "K4me3-R8me2a" histone marks, and more recently the non-canonical bivalent H3 "K4me3-K9me3/2" marks as well. As a robust Spin1-interacting cofactor, Spindoc has been identified to enhance the binding of Spin1 to its substrate histone marks, thereby modulating the downstream signaling; However, the physiological role of Spindoc in germline development is unknown. We generated two Spindoc knockout mouse models through CRISPR/Cas9 strategy, which revealed that Spindoc is specifically required for haploid spermatid development, but not essential for meiotic divisions in spermatocytes. This study unveiled a new epigenetic player that participates in haploid germline development.
Collapse
Affiliation(s)
- Xue Jiang
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China
| | - Xiaoli Zhu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China
| | - Yu Cheng
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China
| | - Muhammad Azhar
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China
| | - Xuemei Xing
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China
| | - Wenqing Li
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China
| | - Yuzhu Cao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China.
| | - Qinghua Shi
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China.
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, People's Republic of China.
| |
Collapse
|
24
|
Weinhouse C. The roles of inducible chromatin and transcriptional memory in cellular defense system responses to redox-active pollutants. Free Radic Biol Med 2021; 170:85-108. [PMID: 33789123 PMCID: PMC8382302 DOI: 10.1016/j.freeradbiomed.2021.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022]
Abstract
People are exposed to wide range of redox-active environmental pollutants. Air pollution, heavy metals, pesticides, and endocrine disrupting chemicals can disrupt cellular redox status. Redox-active pollutants in our environment all trigger their own sets of specific cellular responses, but they also activate a common set of general stress responses that buffer the cell against homeostatic insults. These cellular defense system (CDS) pathways include the heat shock response, the oxidative stress response, the hypoxia response, the unfolded protein response, the DNA damage response, and the general stress response mediated by the stress-activated p38 mitogen-activated protein kinase. Over the past two decades, the field of environmental epigenetics has investigated epigenetic responses to environmental pollutants, including redox-active pollutants. Studies of these responses highlight the role of chromatin modifications in controlling the transcriptional response to pollutants and the role of transcriptional memory, often referred to as "epigenetic reprogramming", in predisposing previously exposed individuals to more potent transcriptional responses on secondary challenge. My central thesis in this review is that high dose or chronic exposure to redox-active pollutants leads to transcriptional memories at CDS target genes that influence the cell's ability to mount protective responses. To support this thesis, I will: (1) summarize the known chromatin features required for inducible gene activation; (2) review the known forms of transcriptional memory; (3) discuss the roles of inducible chromatin and transcriptional memory in CDS responses that are activated by redox-active environmental pollutants; and (4) propose a conceptual framework for CDS pathway responsiveness as a readout of total cellular exposure to redox-active pollutants.
Collapse
Affiliation(s)
- Caren Weinhouse
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, 97214, USA.
| |
Collapse
|
25
|
Barrett A, McWhirter R, Taylor SR, Weinreb A, Miller DM, Hammarlund M. A head-to-head comparison of ribodepletion and polyA selection approaches for C. elegans low input RNA-sequencing libraries. G3-GENES GENOMES GENETICS 2021; 11:6226485. [PMID: 33856427 PMCID: PMC8495925 DOI: 10.1093/g3journal/jkab121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
A recent and powerful technique is to obtain transcriptomes from rare cell populations, such as single neurons in Caenorhabditis elegans, by enriching dissociated cells using fluorescent sorting. However, these cell samples often have low yields of RNA that present challenges in library preparation. This can lead to PCR duplicates, noisy gene expression for lowly expressed genes, and other issues that limit endpoint analysis. Furthermore, some common resources, such as sequence-specific kits for removing ribosomal RNA, are not optimized for nonmammalian samples. To advance library construction for such challenging samples, we compared two approaches for building RNAseq libraries from less than 10 nanograms of C. elegans RNA: SMARTSeq V4 (Takara), a widely used kit for selecting poly-adenylated transcripts; and SoLo Ovation (Tecan Genomics), a newly developed ribodepletion-based approach. For ribodepletion, we used a custom kit of 200 probes designed to match C. elegans rRNA gene sequences. We found that SoLo Ovation, in combination with our custom C. elegans probe set for rRNA depletion, detects an expanded set of noncoding RNAs, shows reduced noise in lowly expressed genes, and more accurately counts expression of long genes. The approach described here should be broadly useful for similar efforts to analyze transcriptomics when RNA is limiting.
Collapse
Affiliation(s)
- Alec Barrett
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Rebecca McWhirter
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Seth R Taylor
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Alexis Weinreb
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - David M Miller
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.,Program in Neuroscience, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Marc Hammarlund
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
26
|
Knudsen TB, Fitzpatrick SC, De Abrew KN, Birnbaum LS, Chappelle A, Daston GP, Dolinoy DC, Elder A, Euling S, Faustman EM, Fedinick KP, Franzosa JA, Haggard DE, Haws L, Kleinstreuer NC, Buck Louis GM, Mendrick DL, Rudel R, Saili KS, Schug TT, Tanguay RL, Turley AE, Wetmore BA, White KW, Zurlinden TJ. FutureTox IV Workshop Summary: Predictive Toxicology for Healthy Children. Toxicol Sci 2021; 180:198-211. [PMID: 33555348 PMCID: PMC8041457 DOI: 10.1093/toxsci/kfab013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
FutureTox IV, a Society of Toxicology Contemporary Concepts in Toxicology workshop, was held in November 2018. Building upon FutureTox I, II, and III, this conference focused on the latest science and technology for in vitro profiling and in silico modeling as it relates to predictive developmental and reproductive toxicity (DART). Publicly available high-throughput screening data sets are now available for broad in vitro profiling of bioactivities across large inventories of chemicals. Coupling this vast amount of mechanistic data with a deeper understanding of molecular embryology and post-natal development lays the groundwork for using new approach methodologies (NAMs) to evaluate chemical toxicity, drug efficacy, and safety assessment for embryo-fetal development. NAM is a term recently adopted in reference to any technology, methodology, approach, or combination thereof that can be used to provide information on chemical hazard and risk assessment to avoid the use of intact animals (U.S. Environmental Protection Agency [EPA], Strategic plan to promote the development and implementation of alternative test methods within the tsca program, 2018, https://www.epa.gov/sites/production/files/2018-06/documents/epa_alt_strat_plan_6-20-18_clean_final.pdf). There are challenges to implementing NAMs to evaluate chemicals for developmental toxicity compared with adult toxicity. This forum article reviews the 2018 workshop activities, highlighting challenges and opportunities for applying NAMs for adverse pregnancy outcomes (eg, preterm labor, malformations, low birth weight) as well as disorders manifesting postnatally (eg, neurodevelopmental impairment, breast cancer, cardiovascular disease, fertility). DART is an important concern for different regulatory statutes and test guidelines. Leveraging advancements in such approaches and the accompanying efficiencies to detecting potential hazards to human development are the unifying concepts toward implementing NAMs in DART testing. Although use of NAMs for higher level regulatory decision making is still on the horizon, the conference highlighted novel testing platforms and computational models that cover multiple levels of biological organization, with the unique temporal dynamics of embryonic development, and novel approaches for estimating toxicokinetic parameters essential in supporting in vitro to in vivo extrapolation.
Collapse
Affiliation(s)
- Thomas B Knudsen
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | | | | | - Linda S Birnbaum
- National Institute of Environmental Health Science, NIH, Research Triangle Park, North Carolina, USA
| | - Anne Chappelle
- Chappelle Toxicology Consulting, LLC, Chadds Ford, Pennsylvania, USA
| | | | | | - Alison Elder
- University of Rochester, Rochester, New York, USA
| | - Susan Euling
- U.S. Environmental Protection Agency, Office of Children’s Health Protection, Washington, District of Columbia, USA
| | | | | | - Jill A Franzosa
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Derik E Haggard
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
- Oak Ridge Institute for Science and Education (ORISE);, Texas, USA
| | | | | | | | - Donna L Mendrick
- U.S. Food and Drug Administration, NCTR, Silver Spring, Maryland, USA
| | | | - Katerine S Saili
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Thaddeus T Schug
- National Institute of Environmental Health Science, NIH, Research Triangle Park, North Carolina, USA
| | | | | | - Barbara A Wetmore
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| | - Kimberly W White
- American Chemistry Council, Washington, District of Columbia, USA
| | - Todd J Zurlinden
- U.S. Environmental Protection Agency, ORD, Research Triangle Park, North Carolina, USA
| |
Collapse
|
27
|
Lee JY, Song J, LeBlanc L, Davis I, Kim J, Beck S. Conserved dual-mode gene regulation programs in higher eukaryotes. Nucleic Acids Res 2021; 49:2583-2597. [PMID: 33621342 PMCID: PMC7969006 DOI: 10.1093/nar/gkab108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/21/2020] [Accepted: 02/08/2021] [Indexed: 12/27/2022] Open
Abstract
Recent genomic data analyses have revealed important underlying logics in eukaryotic gene regulation, such as CpG islands (CGIs)-dependent dual-mode gene regulation. In mammals, genes lacking CGIs at their promoters are generally regulated by interconversion between euchromatin and heterochromatin, while genes associated with CGIs constitutively remain as euchromatin. Whether a similar mode of gene regulation exists in non-mammalian species has been unknown. Here, through comparative epigenomic analyses, we demonstrate that the dual-mode gene regulation program is common in various eukaryotes, even in the species lacking CGIs. In cases of vertebrates or plants, we find that genes associated with high methylation level promoters are inactivated by forming heterochromatin and expressed in a context-dependent manner. In contrast, the genes with low methylation level promoters are broadly expressed and remain as euchromatin even when repressed by Polycomb proteins. Furthermore, we show that invertebrate animals lacking DNA methylation, such as fruit flies and nematodes, also have divergence in gene types: some genes are regulated by Polycomb proteins, while others are regulated by heterochromatin formation. Altogether, our study establishes gene type divergence and the resulting dual-mode gene regulation as fundamental features shared in a broad range of higher eukaryotic species.
Collapse
Affiliation(s)
- Jun-Yeong Lee
- Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04609, USA
| | - Jawon Song
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, TX 78758, USA
| | - Lucy LeBlanc
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ian Davis
- Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04609, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Samuel Beck
- Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04609, USA
| |
Collapse
|
28
|
Li H, Cui D, Zheng L, Zhou Y, Gan L, Liu Y, Pan Y, Zhou X, Wan M. Bisphenol A Exposure Disrupts Enamel Formation via EZH2-Mediated H3K27me3. J Dent Res 2021; 100:847-857. [PMID: 33655795 DOI: 10.1177/0022034521995798] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Enamel formation is a serial and complex biological process, during which related genes are expressed progressively in a spatiotemporal manner. This process is vulnerable to environmental cues, resulting in developmental defects of enamel (DDE). However, how environmental factors are biologically integrated during enamel formation is still poorly understood. Here, we investigated the mechanism of DDE elicited by a model endocrine-disrupting chemical, bisphenol A (BPA), in mouse incisors. We show that BPA exposure leads to DDE in mouse incisors, as well as excessive proliferation in dental epithelial stem/progenitor cells. Western blotting, chromatin immunoprecipitation sequencing, and immunofluorescence staining revealed that this effect was accompanied by upregulation of a repressive mark, H3K27me3, in the labial cervical loop of mouse incisors. Perturbation of H3K27me3 methyltransferase EZH2 repressed the level of H3K27me3 and partially attenuated the excessive proliferation in dental epithelial stem/progenitor cells and DDE induced by BPA exposure. Overall, our results demonstrate the essential role of repressive histone modification H3K27me3 in DDE elicited by exposure to an endocrine-disrupting chemical.
Collapse
Affiliation(s)
- H Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - D Cui
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Gan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Pan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - M Wan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Lister-Shimauchi EH, Dinh M, Maddox P, Ahmed S. Gametes deficient for Pot1 telomere binding proteins alter levels of telomeric foci for multiple generations. Commun Biol 2021; 4:158. [PMID: 33542458 PMCID: PMC7862594 DOI: 10.1038/s42003-020-01624-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 12/15/2020] [Indexed: 11/20/2022] Open
Abstract
Deficiency for telomerase results in transgenerational shortening of telomeres. However, telomeres have no known role in transgenerational epigenetic inheritance. C. elegans Protection Of Telomeres 1 (Pot1) proteins form foci at the telomeres of germ cells that disappear at fertilization and gradually accumulate during development. We find that gametes from mutants deficient for Pot1 proteins alter levels of telomeric foci for multiple generations. Gametes from pot-2 mutants give rise to progeny with abundant POT-1::mCherry and mNeonGreen::POT-2 foci throughout development, which persists for six generations. In contrast, gametes from pot-1 mutants or pot-1; pot-2 double mutants induce diminished Pot1 foci for several generations. Deficiency for MET-2, SET-25, or SET-32 methyltransferases, which promote heterochromatin formation, results in gametes that induce diminished Pot1 foci for several generations. We propose that C. elegans POT-1 may interact with H3K9 methyltransferases during pot-2 mutant gametogenesis to induce a persistent form of transgenerational epigenetic inheritance that causes constitutively high levels of heterochromatic Pot1 foci.
Collapse
Affiliation(s)
- Evan H Lister-Shimauchi
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Michael Dinh
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Paul Maddox
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Shawn Ahmed
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
30
|
Lismer A, Siklenka K, Lafleur C, Dumeaux V, Kimmins S. Sperm histone H3 lysine 4 trimethylation is altered in a genetic mouse model of transgenerational epigenetic inheritance. Nucleic Acids Res 2020; 48:11380-11393. [PMID: 33068438 PMCID: PMC7672453 DOI: 10.1093/nar/gkaa712] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/11/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
Advancing the molecular knowledge surrounding fertility and inheritance has become critical given the halving of sperm counts in the last 40 years, and the rise in complex disease which cannot be explained by genetics alone. The connection between both these trends may lie in alterations to the sperm epigenome and occur through environmental exposures. Changes to the sperm epigenome are also associated with health risks across generations such as metabolic disorders and cancer. Thus, it is imperative to identify the epigenetic modifications that escape reprogramming during spermatogenesis and embryogenesis. Here, we aimed to identify the chromatin signature(s) involved in transgenerational phenotypes in our genetic mouse model of epigenetic inheritance that overexpresses the histone demethylase KDM1A in their germ cells. We used sperm-specific chromatin immunoprecipitation followed by in depth sequencing (ChIP-seq), and computational analysis to identify whether differential enrichment of histone H3 lysine 4 trimethylation (H3K4me3), and histone H3 lysine 27 trimethylation (H3K27me3) serve as mechanisms for transgenerational epigenetic inheritance through the paternal germline. Our analysis on the sperm of KDM1A transgenic males revealed specific changes in H3K4me3 enrichment that predominantly occurred independently from bivalent H3K4me3/H3K27me3 regions. Many regions with altered H3K4me3 enrichment in sperm were identified on the paternal allele of the pre-implantation embryo. These findings suggest that sperm H3K4me3 functions in the transmission of non-genetic phenotypes transgenerationally.
Collapse
Affiliation(s)
- Ariane Lismer
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Canada
| | - Keith Siklenka
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Canada
| | - Christine Lafleur
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| | - Vanessa Dumeaux
- PERFORM Center and Department of Biology, Concordia University, Montreal, Canada
| | - Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Canada
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| |
Collapse
|
31
|
Genome-wide identification and transcriptional modulation of histone variants and modification related genes in the low pH-exposed marine rotifer Brachionus koreanus. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 36:100748. [PMID: 33032078 DOI: 10.1016/j.cbd.2020.100748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/05/2020] [Accepted: 09/18/2020] [Indexed: 11/22/2022]
Abstract
Histone modification is considered to be a major epigenetic control mechanism. These modifications (e.g. acetylation, phosphorylation, and methylation) may affect the interaction of histones with DNA and/or regulate DNA-based processes (e.g., recombination, repair, replication, and transcription) and chromatin remodeling complexes. Despite their significance in metazoan life and evolution, few studies have been conducted to identify genes undergoing epigenetic control modification in aquatic invertebrates. In this study, we identified whole core histones (70 total genes) and post-translational modification (PTM) histone genes (63 total genes) in the marine rotifer Brachionus koreanus through whole-genome analysis, and annotated them according to the human nomenclature. Notably, upon comparative analysis of cis-regulatory motif sequences, we found that B. koreanus core histone protein structures were similar to those of mammals. Furthermore, to examine the effect of parental low pH stress on the offspring's epigenetic regulation, we investigated the expression of PTM genes in two generations of B. koreanus exposed to low pH conditions. Given that the B. koreanus genome does not possess DNA methyltransferase 1 and 3 genes, we concluded that histone genes could be involved as an important epigenetic mechanism in B. koreanus. Therefore, the histone-associated genes identified in this study could be useful for ecotoxicological studies and facilitate the application of chromatin immunoprecipitation sequencing using high-throughput DNA sequencing based on the genome-wide identification of transcription factor binding sites in rotifers.
Collapse
|
32
|
Wamucho A, Heffley A, Tsyusko OV. Epigenetic effects induced by silver nanoparticles in Caenorhabditis elegans after multigenerational exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 725:138523. [PMID: 32305644 PMCID: PMC11370278 DOI: 10.1016/j.scitotenv.2020.138523] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/02/2020] [Accepted: 04/05/2020] [Indexed: 06/11/2023]
Abstract
Multigenerational effects of silver nanoparticles (Ag-NPs) on reproduction of the soil nematode Caenorhabditis elegans have been observed previously. However, mechanisms of this reproductive sensitivity are unknown. Here we examine whether epigenetic changes occur as a result of multigenerational exposure to Ag-NPs and whether such modifications can be inherited by unexposed generations. Changes at histone methylation markers, histone 3 lysine 4 dimethylation (H3K4me2) and histone 3 lysine 9 trimethylation (H3K9me3), known to affect reproduction, as well as changes in the expression of the genes encoding demethylases and methyltransferases associated with the selected markers, were investigated. We exposed C. elegans at EC30 to AgNO3, pristine Ag-NPs, and its environmentally transformed product, sulfidized Ag-NPs (sAg-NPs). Histone methylation levels at H3K4me2 increase in response to pristine Ag-NP exposure and did not recover after rescue from the exposure, suggesting transgenerational inheritance. Compared to pristine Ag-NPs, exposure to transformed sAg-NPs significantly decreased H3K4me2 and H3K9me3 levels. These changes in the histone methylation were also supported by expression of spr-5 and jmjd-2 (H3K4me2 and H3K9me3 demethylases, respectively) and set-30 (H3K4me2 methyltransferase). Our study demonstrates that multigenerational exposure to Ag-NPs induces epigenetic changes that are inherited by unexposed offspring. However, environmental transformations of Ag-NPs may also reduce toxicity via epigenetic mechanisms, such as changes at histone methylation.
Collapse
Affiliation(s)
- Anye Wamucho
- Department of Plant and Soil Sciences, University of Kentucky, 1100 S. Limestone St., Lexington, KY 40546, USA; Department of Toxicology and Cancer Biology, University of Kentucky, 1095 V.A. Drive, Lexington, KY 40536, USA
| | - Allison Heffley
- Department of Plant and Soil Sciences, University of Kentucky, 1100 S. Limestone St., Lexington, KY 40546, USA; Miami University, 501 E. High St., Oxford, OH 45056, USA
| | - Olga V Tsyusko
- Department of Plant and Soil Sciences, University of Kentucky, 1100 S. Limestone St., Lexington, KY 40546, USA; Department of Toxicology and Cancer Biology, University of Kentucky, 1095 V.A. Drive, Lexington, KY 40536, USA.
| |
Collapse
|
33
|
Perera BP, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:176-192. [PMID: 31177562 PMCID: PMC7252203 DOI: 10.1002/em.22311] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 05/02/2023]
Abstract
The genetic material of every organism exists within the context of regulatory networks that govern gene expression, collectively called the epigenome. Epigenetics has taken center stage in the study of diseases such as cancer and diabetes, but its integration into the field of environmental health is still emerging. As the Environmental Mutagenesis and Genomics Society (EMGS) celebrates its 50th Anniversary this year, we have come together to review and summarize the seminal advances in the field of environmental epigenomics. Specifically, we focus on the role epigenetics may play in multigenerational and transgenerational transmission of environmentally induced health effects. We also summarize state of the art techniques for evaluating the epigenome, environmental epigenetic analysis, and the emerging field of epigenome editing. Finally, we evaluate transposon epigenetics as they relate to environmental exposures and explore the role of noncoding RNA as biomarkers of environmental exposures. Although the field has advanced over the past several decades, including being recognized by EMGS with its own Special Interest Group, recently renamed Epigenomics, we are excited about the opportunities for environmental epigenetic science in the next 50 years. Environ. Mol. Mutagen. 61:176-192, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bambarendage P.U. Perera
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Christopher Faulk
- Department of Animal Sciences, University of Minnesota, St. Paul, Minnesota
| | - Laurie K. Svoboda
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Correspondence to: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.
| |
Collapse
|
34
|
Casier K, Boivin A, Carré C, Teysset L. Environmentally-Induced Transgenerational Epigenetic Inheritance: Implication of PIWI Interacting RNAs. Cells 2019; 8:cells8091108. [PMID: 31546882 PMCID: PMC6770481 DOI: 10.3390/cells8091108] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Environmentally-induced transgenerational epigenetic inheritance is an emerging field. The understanding of associated epigenetic mechanisms is currently in progress with open questions still remaining. In this review, we present an overview of the knowledge of environmentally-induced transgenerational inheritance and associated epigenetic mechanisms, mainly in animals. The second part focuses on the role of PIWI-interacting RNAs (piRNAs), a class of small RNAs involved in the maintenance of the germline genome, in epigenetic memory to put into perspective cases of environmentally-induced transgenerational inheritance involving piRNA production. Finally, the last part addresses how genomes are facing production of new piRNAs, and from a broader perspective, how this process might have consequences on evolution and on sporadic disease development.
Collapse
Affiliation(s)
- Karine Casier
- Transgenerational Epigenetics & small RNA Biology, Sorbonne Université, CNRS, Laboratoire Biologie du Développement, Institut de Biologie Paris-Seine, UMR7622, 75005 Paris, France.
| | - Antoine Boivin
- Transgenerational Epigenetics & small RNA Biology, Sorbonne Université, CNRS, Laboratoire Biologie du Développement, Institut de Biologie Paris-Seine, UMR7622, 75005 Paris, France.
| | - Clément Carré
- Transgenerational Epigenetics & small RNA Biology, Sorbonne Université, CNRS, Laboratoire Biologie du Développement, Institut de Biologie Paris-Seine, UMR7622, 75005 Paris, France.
| | - Laure Teysset
- Transgenerational Epigenetics & small RNA Biology, Sorbonne Université, CNRS, Laboratoire Biologie du Développement, Institut de Biologie Paris-Seine, UMR7622, 75005 Paris, France.
| |
Collapse
|
35
|
Advances in epigenetics link genetics to the environment and disease. Nature 2019; 571:489-499. [DOI: 10.1038/s41586-019-1411-0] [Citation(s) in RCA: 566] [Impact Index Per Article: 94.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
|
36
|
Histone Methylation and Memory of Environmental Stress. Cells 2019; 8:cells8040339. [PMID: 30974922 PMCID: PMC6523599 DOI: 10.3390/cells8040339] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023] Open
Abstract
Cellular adaptation to environmental stress relies on a wide range of tightly controlled regulatory mechanisms, including transcription. Changes in chromatin structure and organization accompany the transcriptional response to stress, and in some cases, can impart memory of stress exposure to subsequent generations through mechanisms of epigenetic inheritance. In the budding yeast Saccharomyces cerevisiae, histone post-translational modifications, and in particular histone methylation, have been shown to confer transcriptional memory of exposure to environmental stress conditions through mitotic divisions. Recent evidence from Caenorhabditis elegans also implicates histone methylation in transgenerational inheritance of stress responses, suggesting a more widely conserved role in epigenetic memory.
Collapse
|
37
|
An H3K27me3 demethylase-HSFA2 regulatory loop orchestrates transgenerational thermomemory in Arabidopsis. Cell Res 2019; 29:379-390. [PMID: 30778176 PMCID: PMC6796840 DOI: 10.1038/s41422-019-0145-8] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/22/2019] [Indexed: 01/18/2023] Open
Abstract
Global warming has profound effects on plant growth and fitness. Plants have evolved sophisticated epigenetic machinery to respond quickly to heat, and exhibit transgenerational memory of the heat-induced release of post-transcriptional gene silencing (PTGS). However, how thermomemory is transmitted to progeny and the physiological relevance are elusive. Here we show that heat-induced HEAT SHOCK TRANSCRIPTION FACTOR A2 (HSFA2) directly activates the H3K27me3 demethylase RELATIVE OF EARLY FLOWERING 6 (REF6), which in turn derepresses HSFA2. REF6 and HSFA2 establish a heritable feedback loop, and activate an E3 ubiquitin ligase, SUPPRESSOR OF GENE SILENCING 3 (SGS3)-INTERACTING PROTEIN 1 (SGIP1). SGIP1-mediated SGS3 degradation leads to inhibited biosynthesis of trans-acting siRNA (tasiRNA). The REF6-HSFA2 loop and reduced tasiRNA converge to release HEAT-INDUCED TAS1 TARGET 5 (HTT5), which drives early flowering but attenuates immunity. Thus, heat induces transmitted phenotypes via a coordinated epigenetic network involving histone demethylases, transcription factors, and tasiRNAs, ensuring reproductive success and transgenerational stress adaptation.
Collapse
|
38
|
Shu L, Meng Q, Diamante G, Tsai B, Chen YW, Mikhail A, Luk H, Ritz B, Allard P, Yang X. Prenatal Bisphenol A Exposure in Mice Induces Multitissue Multiomics Disruptions Linking to Cardiometabolic Disorders. Endocrinology 2019; 160:409-429. [PMID: 30566610 PMCID: PMC6349005 DOI: 10.1210/en.2018-00817] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022]
Abstract
The health impacts of endocrine-disrupting chemicals (EDCs) remain debated, and their tissue and molecular targets are poorly understood. In this study, we leveraged systems biology approaches to assess the target tissues, molecular pathways, and gene regulatory networks associated with prenatal exposure to the model EDC bisphenol A (BPA). Prenatal BPA exposure at 5 mg/kg/d, a dose below most reported no-observed-adverse-effect levels, led to tens to thousands of transcriptomic and methylomic alterations in the adipose, hypothalamus, and liver tissues in male offspring in mice, with cross-tissue perturbations in lipid metabolism as well as tissue-specific alterations in histone subunits, glucose metabolism, and extracellular matrix. Network modeling prioritized main molecular targets of BPA, including Pparg, Hnf4a, Esr1, Srebf1, and Fasn as well as numerous less studied targets such as Cyp51 and long noncoding RNAs across tissues, Fa2h in hypothalamus, and Nfya in adipose tissue. Lastly, integrative analyses identified the association of BPA molecular signatures with cardiometabolic phenotypes in mouse and human. Our multitissue, multiomics investigation provides strong evidence that BPA perturbs diverse molecular networks in central and peripheral tissues and offers insights into the molecular targets that link BPA to human cardiometabolic disorders.
Collapse
Affiliation(s)
- Le Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
| | - Qingying Meng
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Brandon Tsai
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Yen-Wei Chen
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
| | - Andrew Mikhail
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Helen Luk
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, California
| | - Patrick Allard
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, California
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
39
|
Perez MF, Lehner B. Intergenerational and transgenerational epigenetic inheritance in animals. Nat Cell Biol 2019; 21:143-151. [PMID: 30602724 DOI: 10.1038/s41556-018-0242-9] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Animals transmit not only DNA but also other molecules, such as RNA, proteins and metabolites, to their progeny via gametes. It is currently unclear to what extent these molecules convey information between generations and whether this information changes according to their physiological state and environment. Here, we review recent work on the molecular mechanisms by which 'epigenetic' information is transmitted between generations over different timescales, and the importance of this information for development and physiology.
Collapse
Affiliation(s)
- Marcos Francisco Perez
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ben Lehner
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain. .,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
40
|
Govindarajan D, Chatterjee C, Shakambari G, Varalakshmi P, Jayakumar K, Balasubramaniem A. Oxidative stress response, epigenetic and behavioral alterations in Caenorhabditis elegans exposed to organophosphorus pesticide quinalphos. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2019. [DOI: 10.1016/j.bcab.2019.01.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Camacho J, Allard P. Histone Modifications: Epigenetic Mediators of Environmental Exposure Memory. Epigenet Insights 2018; 11:2516865718803641. [PMID: 30443644 PMCID: PMC6233965 DOI: 10.1177/2516865718803641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/03/2018] [Indexed: 11/16/2022] Open
Abstract
How organisms retain a memory of ancestral environmental exposure is a phenomenon that is still poorly understood. Recently published work by our group and others, regarding environmentally induced transgenerational effects, have identified an array of mechanisms, with a particular focus on histone modifications, that shed some light on the underlying epigenetic processes driving long-term generational effects.
Collapse
Affiliation(s)
- Jessica Camacho
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Patrick Allard
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
42
|
Chatterjee N, Gim J, Choi J. Epigenetic profiling to environmental stressors in model and non-model organisms: Ecotoxicology perspective. ENVIRONMENTAL HEALTH AND TOXICOLOGY 2018; 33:e2018015-0. [PMID: 30286591 PMCID: PMC6182246 DOI: 10.5620/eht.e2018015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/19/2018] [Indexed: 05/16/2023]
Abstract
Epigenetics, potentially heritable changes in genome function that occur without alterations to DNA sequence, is an important but understudied component of ecotoxicology studies. A wide spectrum of environmental challenge, such as temperature, stress, diet, toxic chemicals, are known to impact on epigenetic regulatory mechanisms. Although the role of epigenetic factors in certain biological processes, such as tumourigenesis, has been heavily investigated, in ecotoxicology field, epigenetics still have attracted little attention. In ecotoxicology, potential role of epigenetics in multi- and transgenerational phenomenon to environmental stressors needs to be unrevealed. Natural variation in the epigenetic profiles of species in responses to environmental stressors, nature of dose-response relationships for epigenetic effects, and how to incorporate this information into ecological risk assessment should also require attentions. In this review, we presented the available information on epigenetics in ecotoxicological context. For this, we have conducted a systemic review on epigenetic profiling in response to environmental stressors, mostly chemical exposure, in model organisms, as well as, in ecotoxicologically relevant wildlife species.
Collapse
Affiliation(s)
- Nivedita Chatterjee
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdaero, Dondaemun-gu, Seoul 02504, Republic of Korea
| | - Jiwan Gim
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdaero, Dondaemun-gu, Seoul 02504, Republic of Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdaero, Dondaemun-gu, Seoul 02504, Republic of Korea
| |
Collapse
|
43
|
Weinhouse C, Truong L, Meyer JN, Allard P. Caenorhabditis elegans as an emerging model system in environmental epigenetics. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:560-575. [PMID: 30091255 PMCID: PMC6113102 DOI: 10.1002/em.22203] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/13/2018] [Accepted: 04/19/2018] [Indexed: 05/19/2023]
Abstract
The roundworm Caenorhabitis elegans has been an established model organism for the study of genetics and developmental biology, including studies of transcriptional regulation, since the 1970s. This model organism has continued to be used as a classical model system as the field of transcriptional regulation has expanded to include scientific advances in epigenetics and chromatin biology. In the last several decades, C. elegans has emerged as a powerful model for environmental toxicology, particularly for the study of chemical genotoxicity. Here, we outline the utility and applicability of C. elegans as a powerful model organism for mechanistic studies of environmental influences on the epigenome. Our goal in this article is to inform the field of environmental epigenetics of the strengths and limitations of the well-established C. elegans model organism as an emerging model for medium-throughput, in vivo exploration of the role of exogenous chemical stimuli in transcriptional regulation, developmental epigenetic reprogramming, and epigenetic memory and inheritance. As the field of environmental epigenetics matures, and research begins to map mechanisms underlying observed associations, new toolkits and model systems, particularly manipulable, scalable in vivo systems that accurately model human transcriptional regulatory circuits, will provide an essential experimental bridge between in vitro biochemical experiments and mammalian model systems. Environ. Mol. Mutagen. 59:560-575, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Caren Weinhouse
- Duke Global Health Institute, Duke University, Durham, North Carolina
- Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Lisa Truong
- UCLA Human Genetics and Genomic Analysis Training Program, University of California, Los Angeles; Los Angeles, California
| | - Joel N. Meyer
- Duke Global Health Institute, Duke University, Durham, North Carolina
- Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Patrick Allard
- Institute for Society and Genetics, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|